Preprint
Article

This version is not peer-reviewed.

In Silico Potential Antiviral Activity of Polyphenols From Plants Used in Traditional Medicine in La Libertad Región, Peru

Submitted:

22 May 2023

Posted:

24 May 2023

You are already at the latest version

Abstract
Traditional medicine is one of the main bases for studying and discovering natural sources of phytometabolites with antiviral properties. This research aims to demonstrate that the medicinal plants used as a treatment of viral diseases in the La Libertad region have, in fact, antiviral ac-tivity. The study evaluated the ethnobotany of the 8 most widely used medicinal plants in the region (Azadirachta indica A. Juss. “paraíso”, Caesalpinia spinosa (Molina) Kuntze “tara”, Citrus limon (L.) Osbeck “limón”, Clinopodium pulchellum (Kunth) Govaerts “panizara”, Cordia lutea Lam. “overo”, Ocimum basilicum L. “albahaca”, Schinus molle L. “molle”, and Taraxacum campylodes G.E. Haglund “diente de león”). The phytometabolites responsible for the antiviral activity were identified by LC-MS and evaluated in silico against the viral proteins NS2B/NS3 (DENV-2), NS5B (HCV), and ICP27 (HSV-1) using molecular docking in Autodock Vina and UCSF Chimera. The presence of five polyphenols (chlorogenic acid, gallic acid, caffeic acid, rosmarinic acid, and rutin) was found and, in the in silico test, the antiviral activity of chlorogenic acid stood out against DENV-2 and HCV, rutin against HCV and HSV-1, rosmarinic acid against DENV-2 and HCV. Therefore, it is verified that the medicinal plants studied have antiviral activity, which supports their use in traditional medicine
Keywords: 
;  ;  ;  ;  

1. Introduction

Synthetic drugs often cause adverse effects and are harmful when the treatment lasts for a prolonged period [1]. On the contrary, natural products regulate the damaging side effects and relieve the harm of various therapies. Moreover, natural products have been used as a source for drug design and development [2]. All these factors make them a possible choice when looking for an efficient treatment [2,3].
In times of health crisis, due to the ineffectiveness of therapies and the lack of medicines to combat a disease, traditional knowledge about medicinal plants and herbs plays an important role [4] During the recent COVID-19 pandemic, this fact was highlighted [5].
As the current pandemic underscored the importance of herbal therapies in most countries [6], ethnobotany took relevance. Ethnobotany is the interrelation of human beings with plants. Nevertheless, this interrelation has been lost multiple times [7,8]. As stated before, this field can add up new medication sources and provide other health prevention benefits. Therefore, there is a need to identify suitable and reliable natural compounds that could be used as drugs [9].
Medicinal plants with potential molecules for ethnobotanical use must be researched and selected with a disease treatment approach. Also, they must comply with traditional pharmacopeias [10], ethnopharmacological evaluations, and therapeutic efficacy reports [11,12]. Likewise, the family of the plant species is not only an ethnomedical indicator but also a key to giving value to plant usage [13,14].
Some plant species are used as traditional medicines to treat diseases of viral origin [15,16,17,18]. Antiviral active principles described include alkaloids, terpenes, polysaccharides, flavonoids, phenolic acids, and steroids [19]. Thus, investigation of new species is necessary to find a source rich in molecules with antiviral activity [20,21]. However, choosing the most biologically active one remains a challenge.
From the COVID-19 pandemic and the global health crisis, not only multiple variants of the Coronavirus (SARS-CoV-2) have emerged, but also latent viruses have reappeared. Some of them, like Hepatitis C Virus (HCV) [22] and Herpes Simplex Virus (HSV) [23], cause the most widespread and harmful chronic infections. Others like the Dengue virus [24] are less harmful. Anyway, they constitute a concern for triggering and increasing the health problem.
On the other hand, the development of new antiviral drugs with efficacy in blocking host-specific functions had stopped. However, the advancement in detection assays, prediction tools, and virtual screening [25], brought the aim back [26]. Therefore, there is a need to reinvestigate medicinal plants and herbs using modern techniques and methods [27].
The perspectives of the study give a vision of the most frequently used plants in traditional medicine. Also, it describes the most relevant species indicated when conditions or symptoms correspond to a disease of viral origin.
The research work carried out aims to demonstrate that the medicinal plants used to treat viral diseases in the La Libertad region have, in fact, antiviral activity.

2. Results and Discussion

2.1. Taxonomic and Ethnobotany of Medicinal Plants

The results of the ethnobotanical study indicate the eight plants that are used and sold the most as natural treatments for viral diseases. Table 1 presents the collected information about their usage and preparation. Also, it shows the Herbarium Truxillense (HUT) code that demonstrates their taxonomical validation.
Figure 1. Most used medicinal plants for viral deases in La Libertad region: Azadirachta indica (A), Caesalpinia spinosa (B), Citrus limon (C), Clinopodium pulchellum (D), Cordia lutea (E), Ocimum basilicum (F), Schinus molle (G), Taraxacum campylodes (H).
Figure 1. Most used medicinal plants for viral deases in La Libertad region: Azadirachta indica (A), Caesalpinia spinosa (B), Citrus limon (C), Clinopodium pulchellum (D), Cordia lutea (E), Ocimum basilicum (F), Schinus molle (G), Taraxacum campylodes (H).
Preprints 74381 g001
Herbalists from local markets played an essential role in the gathering of information. They provided traditional knowledge about plants that people buy, prepare, and use to treat viral diseases. Interestingly, the eight species are used as infusion or decoction.
The most common viral affections treated with these plants are hepatic, dermatological, and mosquito-borne. In line with our data, the bibliography describes these and related plant species with biological activity against a hepatic virus (hepatitis), a dermatological virus (herpes simplex), and a mosquito-borne virus (dengue), as summarized in Table 2.
Literature shows that Azadirachta indica, Citrus limon, Cordia lutea, Ocinum basiculum, and Taraxum campylodes have evidence of antiviral activity. On the other hand, the antiviral activity reported belongs to plants with the same genre but different species. That is the case of Caesalpinia spinosa with C. pucherrima, C. sappan, and C. crista; Clinopodium pulchellum with C. gracile; and Schinus molle with S. terebinthifolia.
Scientific studies validate traditional medicine knowledge by demonstrating its effectiveness. Therefore, the next step in the study was to identify the molecules and phytochemicals responsible for the antiviral effect attributed to these eight plants.

2.2. Antiviral Activity of Phytometabolites Polyphenolics of Medicinal Plants

To identify possible antiviral compounds, the LC-MS/MS method was used. The presence of only five molecules was reported, as stated in Table 3.
The identified phytometabolites are polyphenols. Specifically, one trihydroxy benzoic acid: gallic acid (1); three hydroxycinnamic acids: chlorogenic acid (2), caffeic acid (3), and rosmarinic acid (5); and one flavonol glycoside: rutin (4).
Polyphenols are recognized for their antiviral potential [50,51,52]. Their presence in plants and herbs is extensive [53]; and due to their diverse biological and pharmacological activity, are indicated as therapeutic sources with medicinal and pharmaceutical applications [54,55]. Several studies demonstrate that these phytochemicals have antiviral activity [4,56,57,58]. The evidence includes not only laboratory in vitro and in vivo studies but also clinical trials [57].
Figure 2. Chemical structures of polyphenols identified in medicinal plants from La Libertad region.
Figure 2. Chemical structures of polyphenols identified in medicinal plants from La Libertad region.
Preprints 74381 g002
Due to what has been described, Azadirachta indica, Caesalpinia spinosa, Citrus limon, Clinopodium pulchellum, Cordia lutea, Ocimun basilicum, Schinus molle, and Taraxacum compylodes are considered possible natural antiviral agents because they contain polyphenol-type molecules with active characteristics against several types of viruses. These results confirm what is described in the literature, endorsing the effectiveness of their usage.

2.3. Molecular Docking Analysis

Five polyphenolic compounds found in the most consumed medicinal plants in traditional medicine in the La Libertad region were evaluated in silico using molecular docking in Autodock Vina. Through this program, it is possible to predict the binding affinities and molecular interactions between the compounds and the viral targets. Table 4 shows the results.
In the molecular docking carried out, the affinity between the identified polyphenols and the domain of the following viral proteins was evaluated: NS2B/NS3 (PDB: 2FOM) for DENV-2, NS5B (PDB: 4EO6) for HCV and ICP27 (PDB: 4YXP). for HSV-1.
In the case of DENV-2, several studies have NS2B/NS3 protease as the evaluated molecular target [59,60]. NS2B is a required cofactor of the NS3 serine protease [61]. Together with a cellular protease, this complex cleaves the viral precursor polyprotein into functional viral proteins [62]. Thus, the formation of this complex is essential for the virus replication process. The ligand-protein binding reported by the compounds and the NS2B/NS3 protease ranged from -8.6 kcal/mol to -5.7 kcal/mol. The higher the affinity to the macromolecular target, the lower the binding energy value [63]. Under this tenor, chlorogenic acid showed the highest binding affinity, while gallic acid had the lowest. The results obtained are consistent with those found for other compounds with antiviral potential for DENV-2 [64].
Hydrogen bonding and hydrophobic protein-ligand interactions are also determining factors for molecular docking values [65]. Chlorogenic acid achieved its stability with residues showing six hydrophobic interactions with LEU, 76, TRP83, LEU85, LEU149, VAL 154, ILE123, two anionic interactions with LYS 73, LYS74, three polar interactions with THR118, THR120, and ASN152. Likewise, rutin showed three hydrophobic interactions with VAL72, VAL154, and VAL155, five polar interactions with TRH118, ASN119, THR120, HIS51, and ASN152, two anionic interactions with LYS 73, and LYS74 and one ionic interaction with ASP75.
Figure 3. 2D docked complexes of the molecular interaction between NS2B/NS3 (PDB: 2FOM) domain and chlorogenic acid (A), rutin (B), gallic acid (C), rosmarinic acid (D), and caffeic acid (E).
Figure 3. 2D docked complexes of the molecular interaction between NS2B/NS3 (PDB: 2FOM) domain and chlorogenic acid (A), rutin (B), gallic acid (C), rosmarinic acid (D), and caffeic acid (E).
Preprints 74381 g003
Gallic acid also presented interactions with residues. Six hydrophobic interactions with VAL147, LEU149, ALA164, ILE165, ALA166, LEU76, two polar interactions with ASN152, ASN167 and one anionic interaction with LYS74. Similarly, rosmarinic acid achieved its stability with the interaction of the residues, nine hydrophobic interactions were shown with TRP89, LEU85, TRP83, LEU76, MET49, LEU149, ALA164, VAL147, and VAL146, two ionic interactions with GLU86, GLU88, one interaction polar and anionic with ASN152 and LYS74 respectively.
Likewise, caffeic acid reported hydrophobic interactions through six unions with the residues LEU85, TRP83, VAL146, VAL147, LEU149, and LEU76, an anionic interaction and a polar one with LYS74 and ASN152 respectively.
In agreement with previous studies [66], the interactions found are close to residues LEU76, LEU149, TRP83, and ASN152 stabilizing the active site of DENV-2.
Notably, the chlorogenic acid-NS2B/NS3 interaction had the shortest distance (2.035 Å/2.183 Å RMSD) to a hydrogen bond between the ligand and the target protein (Figure 4). Another remarkable compound for this aspect is rosmarinic acid, whose distance was 2.116 Å/2.179 Å RMSD (Figure 5).
The non-structural protein NS5B is the core of HCV replication [67]. It is an RNA-dependent RNA polymerase (RdRp) that, when interacting with other proteins, promotes the formation of the viral RNA replication complex [68]. Since blocking the function of NS5B can inhibit HCV replication [69], this polymerase is a widely evaluated target. In this investigation, ligand-protein binding ranged from -9.9 kcal/mol to -5.9 kcal/mol. Rutin stood out for its high binding affinity, while gallic acid showed the lowest affinity. Previous studies on the anti-HCV activity of phenolic compounds and flavonoids [70,71,72] support our results. Furthermore, the binding energy value of rutin, chlorogenic acid, and rosmarinic are similar to potential antiviral compounds reported previously [73].
Some interactions with the residues stabilize polyphenols in the 2D simulation of the complexes formed (Figure 6).
Chlorogenic acid showed three hydrophobic interactions with VAL144, PRO404, and VAL405, four polar interactions with ASN142, THR403, SER407, and ASN411, and two ionic interactions with GLU143 and GLU398. In turn, rutin reported eight hydrophobic interactions with ALA450, CYS451, PHE162, ILE160, TYR555, ILE560, TYR561, MET139, two ionic interactions with ASP444 and ASP559, two anionic interactions with LYS 141 and ARG168, and one polar interaction with GLN446. Gallic acid presented hydrophobic interactions with residues VAL161, ILE160, LEU159, CYS223, and PHE224, two anionic interactions with ARG158 and ARG48, two polar interactions with SER282 and SER226, and one ionic interaction with ASP225. Likewise, rosmarinic acid showed six hydrophobic interactions with ALA97, PRO93, ILE560, ILE555, ILE160, and PHE162, two polar interactions with SER96 and SER556, and one ionic and anionic interaction with ASP559 and LYS141 respectively. Similarly, the interactions of the residues with caffeic acid showed four hydrophobic bonds with PRO404, VAL405, ALA39, and TRP397, three polar interactions with ASN142, SER407, and THR403, two ionic interactions with GLU18 and GLU398, and one anionic interaction with ARG394.
As mentioned, the interactions at the active site are close to the residues LEU419, THR229, and PRO133. This fact was previously reported [49], which reinforces the results found in the present study.
Regarding the hydrophilic bonds, the compounds rutin (2,098 Å RMSD) (Figure 7), chlorogenic acid (1,947 Å RMSD) (Figure 8), and rosmarinic acid (1,936 Å RMSD) (Figure 9) stood out with favorable distances.
Finally, the docking between the identified polyphenols and the ICP7 viral target of herpes simplex 1 (HSV-1) was performed. ICP7 regulates SRPK1 with the goal of changing host mRNA by modulating kinase activity, favoring viral transcripts [74]. In other words, ICP7 mediates host shut-off during HSV-1 infection. Also, this protein can interact with and hijack cellular proteins that function throughout the RNA maturation process [75,76].
When performing molecular docking, ligand-protein binding ranged from -7.8 kcal/mol to -5.6 kcal/mol. Rutin exhibited the highest binding affinity and gallic acid the lowest. These results prove that polyphenols block HSV-1 infection by inhibiting ICP27 synthesis [77,78,79].
In the 2D modeling (Figure 10) the stability of the polyphenols was evidenced by the interactions with nearby residues.
Chlorogenic acid showed nine hydrophobic interactions with ALA467, LEU464, TYR463, PHE462, PHE428, ILE425, ILE422, LEU419, ILE432 and one ionic interaction with ASP424.
Likewise, rutin reported four hydrophobic interactions with ILE495, PRO498, PHE349 and PRO396, four polar interactions with THR491, HIS494, GLN346, ASN389 and THR392 and two anionic interactions ARG345, ARG393.
Gallic acid presented one hydrophobic interaction with LEU436, five anionic interactions with ARG416, ARG417, ARG418, ARG435, ARG439, and two polar interactions with THR451 and ASN438.
In turn, rosmarinic acid showed nine hydrophobic interactions with PRO465, LEU315, TRP312, VAL310, ALA361, TRP362, TRP292, PHE303 and ALA354 and one ionic, anionic and polar interaction with GLU358, ARG309 and SER311 respectively.
Regarding caffeic acid, it highlighted two hydrophobic interactions with ALA354 and VAL343, three ionic interactions with GLU358, GLU347, and GLU351, and an anionic and polar interaction with ARG340 and SER355 respectively.
The interactions in the active site showed close binding to GLU358, TRP392, and ALA354 residues, a fact previously reported in other investigations [80,81]. Regarding the distance to a hydrophilic bond, a favorable distance between the viral protein and rutin (2.033 Å RMSD) stood out (Figure 11).
All the ligand-protein interactions found in this research work are favorable since the binding energy values were negative [82]. Likewise, the distances found between the bonding atoms are relevant and of biological importance [83].
The compounds considered relevant and with potential for drug development are those that present good interaction and stability in the active site [84]. The results of this work indicate that chlorogenic acid (for DENV-2 and HCV), rutin (for HCV and HSV-1), and rosmarinic acid (for DENV-2 and HCV) meet said criteria. Therefore, its use as the main molecule to develop new pharmacological compounds aimed at inhibiting specific proteins caused by infections of viral origin should be further studied.

4. Materials and Methods

4.1. Plant Material

Medicinal plants were bought from herbalists of the wholesale markets “La Hermelinda”, “Mayorista”, and “La Unión”. The eight plant species used were Azadirachta indica A. Juss. “paraíso”, Caesalpinia spinosa (Molina) Kuntze “tara”, Citrus limon (L.) Osbeck “limón”, Clinopodium pulchellum (Kunth) Govaerts “panizara”, Cordia lutea Lam. “overo”, Ocimum basilicum L. “albahaca”, Schinus molle L. “molle”, and Taraxacum campylodes G.E. Haglund “diente de león”. The aerial parts of medicinal plants were used and plant organs such as stems and secondary branches were excluded.

4.2. Selection, Collection, and Stabilization of Plant Drugs

For the selection of medicinal plants, the traditional medicine of the region La Libertad was considered. The medicinal plants were collected in packages of approximately 0,5 kg of plant material. Subsequently, a complete specimen of each medicinal plant was taken to the Herbarium Truxillense (HUT) for identification and taxonomic classification. The plant material was transported to the Pharmacognosy laboratory of the Faculty of Pharmacy and Biochemistry. They were cleaned with running water; dried at room temperature, and later in an oven at 40 °C; crushed and grinded in a mill (Corona); and finally, stored in airtight jars until later use [85].

4.3. Ethnobotanical Study

The ethnobotanical study was carried out between January and March 2021. The study area was the three most popular markets for the sale of medicinal plants (Hermelinda, Unión, Mayorista). These markets were visited to inform the study's objectives and hold conversations with herbalists, prior to informed consent from each participant. The representative population was twelve people who had knowledge about medicinal plants and sold them on the markets. For the age range, only adults were considered. The research followed the Code of Ethics of the International Society for Ethnobiology (International Society for Ethnobiology. Code of Ethics of the International Society for Ethnobiology (with 2008 additions). 2006; http://www.ethnobiology.net/what-we-do/core-programs/ise-ethics-program/code-of-ethics/code-in-english/) and the protocol was approved by the Ethics Committee of the National University of Trujillo (code N°: PR003P-2022/CEIFYB). A standard questionnaire was applied through structured semi-open interviews in which the plants used to treat some viral diseases were inquired about, taking into account the vernacular name, uses and traditional medicinal properties, forms, and uses of preparation, dose, and administration of the plant used [86,87].

4.4. Identification of Phytometabolites Polyphenolics from Medicinal Plants Referred to in the Literature and Their Antiviral Activity

An extensive literature search (database: Scopus, ScienceDirect, Google scholar) was performed to identify molecules from plant species and some other traditional medicinal plants that have been reported to contain potential antiviral activity. It was compared and a match was made with each plant species under study. From those studies, the effectiveness against the type of virus and active sites of interaction was also collected, and information from the virtual selection was used to identify phytometabolites that can serve as possible clues for the development of antiviral drugs [88,89,90].

4.5. Identification of Polyphenols by UPLC–PDA–ESI–MS/MS

The profiling of the chemical constituents was performed according to [91], using mass spectrometric analysis carried out on a Waters® ACQUITY Xevo® TQ-XS system composed of an ACQUITY® UPLC HSS T3 system and a Xevo® TQ-XS triple-quadrupole tandem mass spectrometer ESI (-ve mode) as the electrospray ionization (ESI) interface. The column used was a C18 100 mm × 2.1 mm column (p.s., 1.7 µm). The concentration was 1 mg/mL for dry extracts, the mobile phases used were 0.1% water in formic acid LC-MS Merck (Germany) (A) and 0.1% acetonitrile LC-MS Merck (Germany) (B) in formic acid in LC-MS Merck (Germany). The gradient was in phase A: 0-10.50 min from 90-78%, 10.5-19 min from 78-50%, 19-23 min from 50-10%, 23-25 min from 10-5%, 25 -26.50 min 5-90%, 26.50-30 min 90-90%, at 300 µL/min flow. The Survey Scan function was used with scanning in Scan Wave MS, scanning was from 50-1500 m/z. The mass detector parameters were 2.5 (kV) capillary voltage, 40 V cone voltage at a gas flow of 150 L/h, desolvation temperature of 480 °C, and collision energy of 20 eV.

4.6. In Silico Antiviral Activity

4.6.1. Ligand preparation

For molecular docking, six molecules previously identified by LC-MS/MS (CID_1794427: chlorogenic acid, CID_5280805: rutin, CID_370: gallic acid, CID_689043: caffeic acid, CID_5281792: rosmarinic acid) with a 3D-structure were used. SDF from the PubChem database (https://pubchem.ncbi.nlm.nih.gov/). Using the Open Babel software, the 3D-SDF structures were converted to the Mol2 format [92]. The energy minimization of the ligands was done using the MMFF94 force field, using the Avogadro software [93].

4.6.2. Protein preparation

Three viral proteins DENV (PBD ID: 2FOM), HCV (PBD ID: 4EO6), and HSV (PBD ID: 4YXP) were selected from 3D structures of the Protein Data Bank RCSB (https: http://www.rcsb.org/search). Proteins were prepared using the UCSF Chimera V 1.16 program by removing ligands, ions, and water molecules, then parameterized by adding polar hydrogens, removing nonpolar hydrogens, and adding Kollman charges [94].

4.6.3. Molecular docking

Molecular docking between selected molecules and proteins was performed using Autodock Vina and UCSF Chimera [95], based on the Lamarckian genetic model. The highest negative scores were identified and the best poses with UCSF Chimera were saved. Grid box was created using the Grid box preparation module with desired dimensions, for PDB ID: 2FOM it was set to center_x = -11.02, center_y = -12.97, center_z = 9.95 and size_x = 25.21, size_y = 23.10, size_z = 22.62, for PDB ID: 4EO6 was set to center_x = -22.02, center_y = 50.32, center_z = 1.18 and size_x = 32.82, size_y = 51, 05, size_z = 34.88 and for PDB ID: 4YXP was set to center_x = -76.93, center_y = 7.79, center_z = 23.25 and size_x = 37.78, size_y = 38.08, size_z = 35,19. Ten AutoDock Vina runs were performed in each scenario for each ligand with a completeness value of eight. The CASTp server was used to find the binding site of viral proteins [96]. The ligand-protein interactions were visualized using the Maestro V 13.1 software [97].

5. Conclusions

Chlorogenic acid, rutin, gallic acid, caffeic acid, and rosmarinic acid were identified in the eight medicinal plants evaluated. In the in silico evaluation, the antiviral activity of chlorogenic acid stood out against DENV-2 and HCV, rutin against HCV and HSV-1, and rosmarinic acid against DENV-2 and HCV. Therefore, Azadirachta indica, Caesalpinia spinosa, Citrus limon, Clinopodium pulchellum, Cordia lutea, Ocimum basilicum, Schinus molle, and Taraxacum campylodes have antiviral activity.

Author Contributions

Conceptualization, J.E.V.-C.; Methodology, M.L.G-Y. and R.W.B.; accomplished the assortment of field data and wrote the initial draft of the manuscript, J.E.V.-C., C.D.R.-A. and M.L.G.-Y.; designed the project and helped in the revision of the manuscript, R.W.B.; helped in data analysis, S.G.R.-R. and E.A.V.-C.; acquired funds for the project J.E.V.-C.; structured the contents, improved this manuscript by reviewing and editing, J.E.V.-C., S.G.R.-R., and E.A.V.-C.; supervised the project, R.W.B., and M.L.G.-Y. All the authors approved the final manuscript after revision. All authors have read and agreed to the published version of the manuscript.

Funding

This research was financed by the Concytec–World Bank Project "Improvement and Expansion of Services of the National System of Science, Technology and Technological Innovation" through the executing unit ProCiencia 8682–PE (Grant N. 07–2018–FONDECYT–BM–PDAEG–UNT) and the APC was financed by the Concytec Project–World Bank

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

To Dr. E. Mantilla-Rodríguez and Dr. O. Caballero-Aquino, Coordinators of the Doctoral Program beneficiary of the funds of the Concytec - World Bank Project, for their collaboration in the research and publication work through the management of the resources necessary for its development and execution.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

References

  1. Lim, S.; Jeong, I.; Cho, J.; Shin, C.; Kim, K.I.; Shim, B.S.; Ko, S.G.; Kim, B. The natural products targeting on allergic rhinitis: from traditional medicine to modern drug discovery. Antioxidants 2021, 10, 1524. [Google Scholar] [CrossRef]
  2. David, B.; Wolfender, J.L.; Dias, D. A. The pharmaceutical industry and natural products: historical status and new trends. Phytochem. Rev. 2015, 14, 299–315. [Google Scholar] [CrossRef]
  3. Bernardini, S.; Tiezzi, A.; Laghezza Masci, V.; Ovidi, E. Natural products for human health: An historical overview of the drug discovery approaches. Nat. Prod. Res. 2018, 32, 1926–1950. [Google Scholar] [CrossRef]
  4. Yasmin, A.R.; Chia, S.L.; Looi, Q.H.; Omar, A.R.; Noordin, M.M.; Ideris, A. Chapter 7 - Herbal extracts as antiviral agents. In Feed Additives; Florou-Paneri, P., Christaki, E., Giannenas, I., Eds.; Academic Press, 2020; pp. 115–132. [CrossRef]
  5. Mehmood, A.; Khan, S.; Khan, S.; Ahmed, S.; Ali, A.; Xue, M.; Ali, L.; Hamza, M.; Munir, A.; Ur, R.S.; Mehmood, K.A.; Hussain, S.A.; Bai, Q. In silico analysis of quranic and prophetic medicinals plants for the treatment of infectious viral diseases including corona virus. Saudi J. Biol. Sci. 2021, 28, 3137–3151. [Google Scholar] [CrossRef]
  6. Sharma, R.K.; Micali, M.; Rana, B.K.; Pellerito, A.; Singla, R.K. Relevance of ayurveda. therapy of holistic application and classification of herbs. In Indian Herbal Medicines: Antioxidant and Antimicrobial Properties; Sharma, R.K., Micali, M., Rana, B.K., Pellerito, A., Singla, R.K., Eds.; SpringerBriefs in Molecular Science; Springer International Publishing: Cham, 2021; pp. 1–29. [CrossRef]
  7. Bermúdez, A.; Oliveira-Miranda, M.A.; Velázquez, D. La investigación etnobotánica sobre plantas medicinales: Una revisión de sus objetivos y enfoques actuales. Interciencia 2005, 30, 453–459. [Google Scholar]
  8. Lalama, A.J.M.; Montes, C.S.; Zaldumbide, V.M.A. Etnobotánica de plantas medicinales en el cantón Tena, para contribuir al conocimiento, conservación y valoración de la diversidad vegetal de la región Amazónica. Dominio Las Cienc. 2016, 2, 26–52. [Google Scholar]
  9. Thanh, L.T.; Andreadakis, Z.; Kumar, A.; Gómez, R.R.; Tollefsen, S.; Saville, M.; Mayhew, S. The Covid-19 vaccine development landscape. Nat. Rev. Drug Discov. 2020, 19, 305–306. [Google Scholar] [CrossRef]
  10. Ford, J.; Gaoue, O.G. Alkaloid-poor plant families, Poaceae and Cyperaceae, are over-utilized for medicine in Hawaiian pharmacopoeia. Econ. Bot. 2017, 71, 123–132. [Google Scholar] [CrossRef]
  11. Kutal, D.H.; Kunwar, R.M.; Uprety, Y.; Adhikari, Y.P.; Bhattarai, S.; Adhikari, B.; Kunwar, L.M.; Bhatt, M.D.; Bussmann, R.W. Selection of medicinal plants for traditional medicines in Nepal. J. Ethnobiol. Ethnomedicine 2021, 17, 59. [Google Scholar] [CrossRef]
  12. Robles, A.D.M.; Cevallos, D.; Gaoue, O.G.; Fadiman, M.G.; Hindle, T. Non-random medicinal plants selection in the Kichwa community of the Ecuadorian amazon. J. Ethnopharmacol. 2020, 246, 112220. [Google Scholar] [CrossRef] [PubMed]
  13. Kutal, D.; Kunwar, R.M.; Baral, K.; Sapkota, P.; Sharma, H.P.; Rimal, B. Factors that influence the plant use knowledge in the middle mountains of Nepal. PLOS ONE 2021, 16, e0246390. [Google Scholar] [CrossRef]
  14. Oliveira, A.C.S.; Junior, N.J.P.B.; Serejo, A.P.M.; Costa, I.S.; Neto, A.C.O.; Godinho, J.W.L.S.; Kzam, P.M.; Amaral, F.M.M. Espécies vegetais de uso popular no tratamento da dor: uma revisão sistemática. Res. Soc. Dev. 2022, 11, e22511225608–e22511225608. [Google Scholar] [CrossRef]
  15. Güçlü, İ.; Yüksel, V. Fitoterapide antiviral bitkiler. Deney. Tıp Araşt. Enstitüsü Derg. 2017, 7, 25–34. [Google Scholar]
  16. Denaro, M.; Smeriglio, A.; Barreca, D.; De Francesco, C.; Occhiuto, C.; Milano, G.; Trombetta, D. Antiviral activity of plants and their isolated bioactive compounds: An update. Phytother. Res. 2020, 34, 742–768. [Google Scholar] [CrossRef]
  17. Huaccho-Rojas, J.; Balladares, A.; Yanac-Tellería, W.; Rodríguez, C.; Villar-López, M. Review of antiviral and immunomodulatory effects of herbal medicine with reference to pandemic COVID-19. Arch. Venez. de Farmacol. y Ter. 2020, 795–807. [Google Scholar]
  18. Akram, M.; Tahir, I.M.; Shah, S.M.A.; Mahmood, Z.; Altaf, A.; Ahmad, K.; Munir, N.; Daniyal, M.; Nasir, S.; Mehboob, H. Antiviral potential of medicinal plants against HIV, HSV, influenza, hpatitis, and coxsackievirus: A systematic review. Phytother. Res. 2018, 32, 811–822. [Google Scholar] [CrossRef]
  19. van de Sand, L.; Bormann, M.; Schmitz, Y.; Heilingloh, C.S.; Witzke, O.; Krawczyk, A. Antiviral active compounds derived from natural sources against herpes simplex viruses. Viruses 2021, 13, 1386. [Google Scholar] [CrossRef]
  20. Mukherjee, P.K. Quality control and evaluation of herbal drugs: evaluating natural products and traditional medicine; Elsevier: Amsterdam, Netherlands, 2019; pp. 599–628. [Google Scholar]
  21. Tida, M.M.A.; Nanjarisoa, O.; Rabearivony, J.; Ranarijaona, H.L.T.; Fenoradosoa, T.A. Ethnobotanical survey of plant species used in traditional medicine in Bekaraoka region, northeastern Madagascar. Int. J. Adv. Res. Publ. 2020, 4, 107–114. [Google Scholar]
  22. Yousaf, T.; Rafique, S.; Wahid, F.; Rehman, S.; Nazir, A.; Rafique, J.; Aslam, K.; Shabir, G.; Shah, S.M. Phytochemical profiling and antiviral activity of Ajuga bracteosa, Ajuga parviflora, Berberis lycium and Citrus lemon against hepatitis C virus. Microb. Pathog. 2018, 118, 154–158. [Google Scholar] [CrossRef]
  23. Krenn, V.; Bosone, C.; Burkard, T.R.; Spanier, J.; Kalinke, U.; Calistri, A.; Salata, C.; Christoff, R.R.; Garcez, P.P.; Mirazimi, A.; Knoblich, J.A. Organoid modeling of zika and herpes simplex virus 1 infections reveals virus-specific responses leading to microcephaly. Cell Stem Cell 2021, 28, 1362–1379. [Google Scholar] [CrossRef] [PubMed]
  24. Yan, G.; Lee, C.K.; Lam, L.T.M.; Yan, B.; Chua, Y.X.; Lim, A.Y.N.; Phang, K.F.; Kew, G.S.; Teng, H.; Ngai, C.H.; Lin, L.; Foo, R.M.; Pada, S.; Ng, L.C.; Tambyah, P.A. Covert COVID-19 and false-positive dengue serology in Singapore. Lancet Infect. Dis. 2020, 20, 536. [Google Scholar] [CrossRef] [PubMed]
  25. Rolta, R.; Yadav, R.; Salaria, D.; Trivedi, S.; Imran, M.; Sourirajan, A.; Baumler, D.J.; Dev, K. In silico screening of hundred phytocompounds of ten medicinal plants as potential inhibitors of nucleocapsid phosphoprotein of Covid-19: An approach to prevent virus assembly. J. Biomol. Struct. Dyn. 2021, 39, 7017–7034. [Google Scholar] [CrossRef] [PubMed]
  26. Martinez, J.P.; Sasse, F.; Brönstrup, M.; Diez, J.; Meyerhans, A. Antiviral drug discovery: Broad-spectrum drugs from nature. Nat. Prod. Rep. 2014, 32, 29–48. [Google Scholar] [CrossRef] [PubMed]
  27. Rani, J.M.J.; Kalaimathi, K.; Vijayakumar, S.; Varatharaju, G.; Karthikeyan, K.; Thiyagarajan, G.; Bhavani, K.; Manogar, P.; Prabhu, S. Anti-viral effectuality of plant polyphenols against mutated dengue protein NS2B47-NS3: A computational exploration. Gene Rep. 2022, 27, 101546. [Google Scholar] [CrossRef]
  28. Wandscheer, C.B.; Duque, J.E.; da Silva, M.A.N.; Fukuyama, Y.; Wohlke, J.L.; Adelmann, J.; Fontana, J.D. Larvicidal action of ethanolic extracts from fruit endocarps of Melia azedarach and Azadirachta indica against the dengue mosquito Aedes aegypti. Toxicon 2004, 44, 829–835. [Google Scholar] [CrossRef] [PubMed]
  29. Dwivedi, V.D.; Bharadwaj, S.; Afroz, S.; Khan, N.; Ansari, M.A.; Yadava, U.; Tripathi, R.C.; Tripathi, I.P.; Mishra, S.K.; Kang, S.G. Anti-dengue infectivity evaluation of bioflavonoid from Azadirachta indica by dengue virus serine protease inhibition. J. Biomol. Struct. Dyn. 2021, 39, 1417–1430. [Google Scholar] [CrossRef]
  30. Chen, X.B.; Liu, X.C.; Zhou, L.; Liu, Z.L. Essential oil composition and larvicidal activity of Clinopodium Gracile (Benth) Matsum (Labiatae) aerial parts against the Aedes Albopictus Mosquito. Trop. J. Pharm. Res. 2013, 12, 799–804. [Google Scholar] [CrossRef]
  31. Murugan, K.; Murugan, P.; Noortheen, A. Larvicidal and repellent potential of Albizzia amara boivin and Ocimum basilicum Linn against dengue vector, Aedes aegypti (Insecta:diptera:culicidae). Bioresour. Technol. 2007, 98, 198–201. [Google Scholar] [CrossRef]
  32. Soonwera, M.; Sittichok, S. Adulticidal activities of Cymbopogon citratus (stapf.) and Eucalyptus globulus (labill.) essential oils and of their synergistic combinations against Aedes Aegypti (L.), Aedes Albopictus (Skuse), and Musca Domestica (L.). Environ. Sci. Pollut. Res. 2020, 27, 20201–20214. [Google Scholar] [CrossRef] [PubMed]
  33. Misra, R.C.; Sharma, S.; Garg, A.; Ghosh, S. Virus-induced gene silencing in sweet basil (Ocimum basilicum). in virus-induced gene silencing in plants: methods and protocols; Courdavault, V., Besseau, S., Eds.; Methods in Molecular Biology; Springer US: New York, NY, 2020; pp. 123–138. [CrossRef]
  34. Kanna, S.U.; Krishnakumar, N. Anti-dengue medicinal plants: A mini review. J. Pharmacogn. Phytochem. 8, 4245–4249.
  35. Procópio, T.F.; Fernandes, K.M.; Pontual, E.V.; Ximenes, R.M.; Oliveira, A.R.C.; Souza, C.S.; Melo, A.M.M.A.; Navarro, D.M.A.F.; Paiva, P.M.G.; Martins, G.F.; Napoleão, T.H. Schinus Terebinthifolius leaf extract causes midgut damage, interfering with survival and development of Aedes Aegypti larvae. PLoS ONE 2015, 10, e0126612. [Google Scholar] [CrossRef] [PubMed]
  36. Tseghai, G.B.; Belay, T.G.; Gebremariam, A.H. Mosquito repellent finishing of cotton using pepper tree (Schinus molle) seed oil extract. J. Text. Sci. Eng. 2019, 9. [Google Scholar]
  37. Lopes, N.; Faccin-Galhardi, L.C.; Espada, S.F.; Pacheco, A.C.; Ricardo, N.M.P.S.; Linhares, R.E.C.; Nozawa, C. Sulfated polysaccharide of Caesalpinia ferrea inhibits herpes simplex virus and poliovirus. Int. J. Biol. Macromol. 2013, 60, 93–99. [Google Scholar] [CrossRef]
  38. Chiang, L.C.; Chiang, W.; Liu, M.C.; Lin, C.C. In vitro antiviral activities of Caesalpinia Pulcherrima and its related flavonoids. J. Antimicrob. Chemother. 2003, 52, 194–198. [Google Scholar] [CrossRef]
  39. Agrawal, P.K.; Agrawal, C.; Blunden, G. Pharmacological significance of hesperidin and hesperetin, two Citrus flavonoids, as promising antiviral compounds for prophylaxis against and combating COVID-19. Nat. Prod. Commun. 2021, 16, 1934578X211042540. [Google Scholar] [CrossRef]
  40. Garber, A.; Barnard, L.; Pickrell, C. Review of whole plant extracts with activity against herpes simplex viruses in vitro and in vivo. J. Evid.-Based Integr. Med. 2021, 26, 2515690X20978394. [Google Scholar] [CrossRef]
  41. Koch, C.; Reichling, J.; Schnitzler, P. Essential oils inhibit replication of herpes simplex virus type 1 and type 2. Bot. Med. Clin. Pract. 2008, 192–197. [Google Scholar] [CrossRef]
  42. Oliveira, M.B.S.; Valentim, I.B.; Rocha, T.S.; Santos, J.C.; Pires, K.S.N.; Tanabe, E.L.L.; Borbely, K.S.C.; Borbely, A.U.; Goulart, M.O.F. Schinus terebenthifolius Raddi extracts: from sunscreen activity toward protection of the placenta to zika virus infection, new uses for a well-known medicinal plant. Ind. Crops Prod. 2020, 152, 112503. [Google Scholar] [CrossRef] [PubMed]
  43. Nocchi, S.R.; Ferreira, L.A.O.; Castro-Hoshino, L.V.; Truiti, M.C.T.; Natali, M.R.M.; Mello, J.C.P.; Baesso, M.L.; Dias, F.B.P.; Nakamura, C.V.; Ueda-Nakamura, T. Development and evaluation of topical formulations that contain hydroethanolic extract from Schinus Terebinthifolia against HSV-1 infection. Braz. J. Pharm. Sci. 2022, 58, e18637. [Google Scholar] [CrossRef]
  44. Nocchi, S.R.; Companhoni, M.V.P.; Mello, J.C.P. de; Filho, B.P.D.; Nakamura, C.V.; Carollo, C.A.; Silva, D.B.; Ueda-Nakamura, T. Antiviral activity of crude hydroethanolic extract from Schinus Terebinthifolia against herpes simplex virus type 1. Planta Med. 2017, 234, 509–518. [Google Scholar] [CrossRef] [PubMed]
  45. Holidah, D.; Dewi, I.P.; Siregar, I.P.A.; Aftiningsih, D. Hepatoprotective effect of Caesalpinia Sappan L. ethanolic extract on alloxan induced diabetic rats: J. Farm. Galen. Galen. J. Pharm. E-J. 2022, 8, 1–9. [Google Scholar] [CrossRef]
  46. Gaurav, S.; Jeyabalan, G.; Anil, A. Antioxidant and skeletal muscle relaxant activity of leaf extract of plant Piper Attenuatum (B. HAM). Indian J. Pharm. Biol. Res. 2021, 9, 8–15. [Google Scholar] [CrossRef]
  47. Ruiz-Reyes, S.G.; Villarreal-La Torre, V.E.; Silva-Correa, C.R.; Sagastegui, G.W.A.; Cruzado-Razco, J.L.; Gamarra-Sánchez, C.D.; Venegas, C.E.A.; Miranda-Leyva, M.; Valdiviezo, C.J.E.; Armando, C.C. Hepatoprotective activity of Cordia lutea Lam flower extracts against paracetamol-induced hepatotoxicity in rats. Pharmacogn. J. 2021, 13, 309–316. [Google Scholar] [CrossRef]
  48. Hayashi, K.; Hayashi, T.; Morita, N.; Niwayama, S. Antiviral activity of an extract of Cordia Salicifolia on herpes simplex virus type 1. Planta Med. 1990, 56, 439–443. [Google Scholar] [CrossRef]
  49. Rehman, S.; Ijaz, B.; Fatima, N.; Muhammad, S.A.; Riazuddin, S. Therapeutic potential of Taraxacum officinale against HCV NS5B polymerase: In vitro and in silico study. Biomed. Pharmacother. 2016, 83, 881–891. [Google Scholar] [CrossRef]
  50. Zhang, Z.J.; Morris-Natschke, S.L.; Cheng, Y.Y.; Lee, K.H.; Li, R.T. Development of anti-influenza agents from natural products. Med. Res. Rev. 2020, 40, 2290–2338. [Google Scholar] [CrossRef]
  51. Kamboj, A.; Saluja, A.K.; Kumar, M.; Atri, P. Antiviral activity of plant polyphenols. J. Pharm. Res. 2012, 5, 2402–2412. [Google Scholar]
  52. Badshah, S.L.; Faisal, S.; Muhammad, A.; Poulson, B.G.; Emwas, A.H.; Jaremko, M. Antiviral activities of flavonoids. Biomed. Pharmacother. 2021, 140, 111596. [Google Scholar] [CrossRef]
  53. Ghasemzadeh, A; Ghasemzadeh, N. Flavonoids and phenolic acids: role and biochemical activity in plants and human. J. Med. Plants Res. 2011, 5. [Google Scholar] [CrossRef]
  54. Kulkarni, K.; Jagtap, G.; Magdum, S. A Comprehensive review on herbal drug standardization. Am. J. PharmTech Res. 2019, 9, 97–122. [Google Scholar] [CrossRef]
  55. Tungmunnithum, D.; Thongboonyou, A.; Pholboon, A.; Yangsabai, A. Flavonoids and other phenolic compounds from medicinal plants for pharmaceutical and medical aspects: An overview. Medicines 2018, 5, 93. [Google Scholar] [CrossRef] [PubMed]
  56. El Sayed, K.A. Natural products as antiviral agents. In Studies in Natural Products Chemistry; Atta-ur-Rahman, Ed.; Bioactive Natural Products (Part E); Elsevier, 2000; Volume 24, pp. 473–572. [CrossRef]
  57. Martin, K.W.; Ernst, E. Antiviral agents from plants and herbs: A systematic review. Antivir. Ther. 2003, 8, 77–90. [Google Scholar] [CrossRef]
  58. Ghildiyal, R.; Prakash, V.; Chaudhary, V.K.; Gupta, V.; Gabrani, R. Phytochemicals as antiviral agents: recent updates. In plant-derived bioactives: production, properties and therapeutic applications; Swamy, M.K., Ed.; Springer: Singapore, 2020; pp. 279–295. [Google Scholar] [CrossRef]
  59. de Sousa, L.R.F.; Wu, H.; Nebo, L.; Fernandes, J.B.; da Silva, M.F.G.F.; Kiefer, W.; Kanitz, M.; Bodem, J.; Diederich, W.E.; Schirmeister, T.; Vieira, P.C. Flavonoids as noncompetitive inhibitors of dengue virus NS2B-NS3 protease: Inhibition kinetics and docking studies. Bioorg. Med. Chem. 2015, 23, 466–470. [Google Scholar] [CrossRef]
  60. Lim, H.; Nguyen, T.T.H.; Kim, N.M.; Park, J.S.; Jang, T.S.; Kim, D. Inhibitory effect of flavonoids against NS2B-NS3 protease of zika virus and their structure activity relationship. Biotechnol. Lett. 2017, 39, 415–421. [Google Scholar] [CrossRef]
  61. Henrich, S.; Feierberg, I.; Wang, T.; Blomberg, N.; Wade, R.C. Comparative binding energy analysis for binding affinity and target selectivity prediction. Proteins Struct. Funct. Bioinforma. 2010, 78, 135–153. [Google Scholar] [CrossRef]
  62. Wu, H.; Bock, S.; Snitko, M.; Berger, T.; Weidner, T.; Holloway, S.; Kanitz, M.; Diederich, W. E.; Steuber, H.; Walter, C.; Hofmann, D.; Weißbrich, B.; Spannaus, R.; Acosta, E.G.; Bartenschlager, R.; Engels, B.; Schirmeister, T.; Bodem, J. Novel dengue virus NS2B/NS3 protease inhibitors. Antimicrob. Agents Chemother. 2015, 59, 1100–1109. [Google Scholar] [CrossRef]
  63. Herowati, R.; Widodo, G.P.; Herowati, R.; Widodo, G.P. Chapter 5: Molecular docking analysis: interaction studies of natural compounds to anti-inflammatory targets; Kandemirli, F., Eds.; IntechOpen, 2017, pp. 63–73. [CrossRef]
  64. Lima, C.S.; Mottin, M.; de Assis, L.R.; Mesquita, N.C.M.R.; Sousa, B.K.P.; Coimbra, L.D.; Santos, K.B.; Zorn, K.M.; Guido, R.V.C.; Ekins, S.; Marques, R.E.; Proença-Modena, J.L.; Oliva, G.; Andrade, C.H.; Regasini, L.O. Flavonoids from Pterogyne Nitens as zika virus NS2B-NS3 protease inhibitors. Bioorganic Chem. 2021, 109, 104719. [Google Scholar] [CrossRef] [PubMed]
  65. Trujillo-Correa, A.I.; Quintero-Gil, D.C.; Diaz-Castillo, F.; Quiñones, W.; Robledo, S.M.; Martinez-Gutierrez, M. In vitro and in silico anti-dengue activity of compounds obtained from Psidium Guajava through Bioprospecting. BMC Complement. Altern. Med. 2019, 19, 298. [Google Scholar] [CrossRef]
  66. Hassandarvish, P.; Rothan, H.A.; Rezaei, S.; Yusof, R.; Abubakar, S.; Zandi, K. In silico study on baicalein and baicalin as inhibitors of dengue virus replication. RSC Adv. 2016, 6, 31235–31247. [Google Scholar] [CrossRef]
  67. Fadl, N.; Salem, T.Z. Hepatitis C genotype 4: A report on resistance-associated substitutions in NS3, NS5A, and NS5B genes. Rev. Med. Virol. 2020, 30, e2120. [Google Scholar] [CrossRef] [PubMed]
  68. Ahmed, H.R.; Waly, N.G.F. M.; El-Baky, R.M.A.; Yahia, R.; Hetta, H.F.; Elsayed, A.M.; Ibrahem, R.A. Distribution of naturally occurring NS5B resistance-associated substitutions in Egyptian patients with chronic hepatitis C. PLOS ONE 2021, 16, e0249770. [Google Scholar] [CrossRef] [PubMed]
  69. Zhou, Z.; Zhang, J.; Zhou, E.; Ren, C.; Wang, J.; Wang, Y. Small molecule NS5B RdRp non-nucleoside inhibitors for the treatment of HCV infection: A medicinal chemistry perspective. Eur. J. Med. Chem. 2022, 240, 114595. [Google Scholar] [CrossRef] [PubMed]
  70. Mustafa, G.; Majid, M.; Ghaffar, A.; Yameen, M.; Samad, H.; Mahrosh, H. Screening and molecular docking of selected phytochemicals against NS5B polymerase of hepatitis C virus. Pak. J. Pharm. Sci. 2020, 33, 2317–2322. [Google Scholar] [CrossRef]
  71. Akher, F.B.; Farrokhzadeh, A.; Ramharack, P.; Shunmugam, L.; van Heerden, F. R.; Soliman, M.E.S. Discovery of novel natural flavonoids as potent antiviral candidates against hepatitis C virus NS5B polymerase. Med. Hypotheses 2019, 132, 109359. [Google Scholar] [CrossRef]
  72. Arabi, I.I.; Mohinuddin, M.; Abir, A.Y.; Koo, S.; Alam, D.M.S. In-silico investigations on the antiviral activity of some polyphenolic derivatives: chemical reactivity descriptors, ADMET, QSAR and molecular docking studies against hepatitis C virus NS5B polymerase. Rochester, NY November 11, 2022. https://papers.ssrn.com/abstract=4267458 (accessed 2023-02-27).
  73. Lakshmi, M.V.; Swapna, T.S.A. Computational study on cosmosiin, an antiviral compound from Memecylon randerianum S.M.Almeida & M.R.Almeida. Med. Plants - Int. J. Phytomedicines Relat. Ind. 2021, 13, 515–523. [Google Scholar] [CrossRef]
  74. Tunnicliffe, R.B.; Hu, W.K.; Wu, M.Y.; Levy, C.; Mould, A.P.; McKenzie, E.A.; Sandri-Goldin, R.M.; Golovanov, A.P. Molecular mechanism of SR protein kinase 1 inhibition by the herpes virus protein ICP27. mBio 2019, 10, e02551–19. [Google Scholar] [CrossRef] [PubMed]
  75. Sandri-Goldin, R.M. The many roles of the highly interactive HSVprotein ICP27, a key regulator of infection. Future Microbiol. 2011, 6, 1261–1277. [Google Scholar] [CrossRef] [PubMed]
  76. Wang, X.; Hennig, T.; Whisnant, A.W.; Erhard, F.; Prusty, B.K.; Friedel, C.C.; Forouzmand, E.; Hu, W.; Erber, L.; Chen, Y.; Sandri-Goldin, R.M.; Dölken, L.; Shi, Y. Herpes simplex virus blocks host transcription termination via the bimodal activities of ICP27. Nat. Commun. 2020, 11, 293. [Google Scholar] [CrossRef] [PubMed]
  77. Borenstein, R.; Hanson, B.A.; Markosyan, R.M.; Gallo, E.S.; Narasipura, S.D.; Bhutta, M.; Shechter, O.; Lurain, N.S.; Cohen, F.S.; Al-Harthi, L.; Nicholson, D.A. Ginkgolic acid inhibits fusion of enveloped viruses. Sci. Rep. 2020, 10, 4746. [Google Scholar] [CrossRef]
  78. Hassan, A.; Abu, H.S.H.; Elghandour, M.M.Y.; Kanth, R.P.R.; Salem, M.Z.M.; Anele, U.Y.; Ranga, R.P.P.; Salem, A.Z.M. Influence of Corymbia Citriodora leaf extract on growth performance, ruminal fermentation, nutrient digestibility, plasma antioxidant activity and faecal bacteria in young calves. Anim. Feed Sci. Technol. 2020, 261, 114394. [Google Scholar] [CrossRef]
  79. Treml, J.; Gazdová, M.; Šmejkal, K.; Šudomová, M.; Kubatka, P.; Hassan, S.T.S. Natural products derived chemicals: Breaking barriers to novel anti-HSV drug development. Viruses 2020, 12, 154. [Google Scholar] [CrossRef]
  80. Yuan, F.; Gao, Z.Q.; Majerciak, V.; Bai, L.; Hu, M.L.; Lin, X.X.; Zheng, Z.M.; Dong, Y.H.; Lan, K. The crystal structure of KSHV ORF57 reveals dimeric active sites important for protein stability and function. PLOS Pathog. 2018, 14, e1007232. [Google Scholar] [CrossRef]
  81. Tunnicliffe, R.B.; Collins, R.F.; Ruiz, N.H.D.; Sandri-Goldin, R.M.; Golovanov, A.P. The ICP27 homology domain of the human cytomegalovirus protein UL69 adopts a dimer of dimers structure. mBio 2018, 9, e01112–18. [Google Scholar] [CrossRef]
  82. Amir-Hassan, A.; Lee, V.S.; Baharuddin, A.; Othman, S.; Xu, Y.; Huang, M.; Yusof, R.; Rahman, N.; Othman, R. Conformational and energy evaluations of novel peptides binding to dengue virus envelope protein. J. Mol. Graph. Model. 2017, 74, 273–287. [Google Scholar] [CrossRef]
  83. Nittinger, E.; Inhester, T.; Bietz, S.; Meyder, A.; Schomburg, K.T.; Lange, G.; Klein, R.; Rarey, M. Large scale analysis of hydrogen bond interaction patterns in protein ligand interfaces. J. Med. Chem. 2017, 60, 4245–4257. [Google Scholar] [CrossRef] [PubMed]
  84. Yadav, R.; Selvaraj, C.; Aarthy, M.; Kumar, P.; Kumar, A.; Singh, S.K.; Giri, R. Investigating into the molecular interactions of flavonoids targeting NS2B-NS3 protease from zika virus through in silico approaches. J. Biomol. Struct. Dyn. 2021, 39, 272–284. [Google Scholar] [CrossRef] [PubMed]
  85. Ruiz, R.S.G.; Venegas, C.E.A.; Valdiviezo, C.J.E.; Ocaña, V.J.P.; Tadeo, H.M.A.V. Características farmacognósticas y cuantificación espectrofotométrica de antocianinas totales del fruto de Prunus serotina subsp. capuli (Cav.) McVaugh (Rosaceae) “capulí.” Arnaldoa 2018, 25, 961–980. [Google Scholar] [CrossRef]
  86. Bussmann, R.W.; Zambrana, P.; Narel, Y.; Sikharulidze, S.; Kikvidze, Z.; Kikodze, D.; Tchelidze, D.; Batsatsashvili, K.; Hart; Robbie, E. Ethnobotany of Samtskhe-Javakheti, Sakartvelo (Republic of Georgia), Caucasus. Indian J. Tradit. Knowl. 2017, 16, 7–24.
  87. Zambrano-Intriago, L.F.; Buenaño-Allauca, M.P.; Mancera-Rodríguez, N.J.; Jiménez-Romero, E. Estudio etnobotánico de plantas medicinales utilizadas por los habitantes del área rural de la Parroquia San Carlos, Quevedo, Ecuador. Univ. Salud 2015, 17, 97–111. [Google Scholar]
  88. Rivero-Segura, N.A.; Gomez-Verjan, J.C. In silico screening of natural products isolated from Mexican herbal medicines against COVID-19. Biomolecules 2021, 11, 216. [Google Scholar] [CrossRef] [PubMed]
  89. Kim, J.; Sohn, S.I.; Sathasivam, R.; Khaskheli, A.J.; Kim, M.C.; Kim, N.S.; Park, S.U. Targeted metabolic and in silico analyses highlight distinct glucosinolates and phenolics signatures in Korean rapeseed cultivars. Plants 2021, 10, 2027. [Google Scholar] [CrossRef]
  90. Mithilesh, S.; Raghunandan, D.; Suresh, P.K. In silico identification of natural compounds from traditional medicine as potential drug leads against SARS-CoV-2 through virtual screening. Proc. Natl. Acad. Sci. India Sect. B Biol. Sci. 2022, 92, 81–87. [Google Scholar] [CrossRef]
  91. El-Zahar, H.; Menze, E.T.; Handoussa, H.; Osman, A.K.; El-Shazly, M.; Mostafa, N.M.; Swilam, N. UPLC-PDA-MS/MS profiling and healing activity of polyphenol rich fraction of Alhagi maurorum against oral ulcer in rats. Plants 2022, 11, 455. [Google Scholar] [CrossRef]
  92. Hanwell, M.D.; Curtis, D.E.; Lonie, D.C.; Vandermeersch, T.; Zurek, E.; Hutchison, G.R. Avogadro: an advanced semantic chemical editor, visualization, and analysis platform. J. Cheminformatics 2012, 4, 17. [Google Scholar] [CrossRef]
  93. Pettersen, E.F.; Goddard, T.D.; Huang, C.C.; Couch, G.S.; Greenblatt, D.M.; Meng, E.C.; Ferrin, T.E. UCSF chimera—A visualization system for exploratory research and analysis. J. Comput. Chem. 2004, 25, 1605–1612. [Google Scholar] [CrossRef] [PubMed]
  94. O’Boyle, N.M.; Banck, M.; James, C.A.; Morley, C.; Vandermeersch, T.; Hutchison, G.R. Open babel: an open chemical toolbox. J. Cheminformatics 2011, 3, 33. [Google Scholar] [CrossRef]
  95. Tian, W.; Chen, C.; Lei, X.; Zhao, J.; Liang, J. CASTp 3.0: Computed atlas of surface topography of proteins. Nucleic Acids Res. 2018, 46, W363–W367. [Google Scholar] [CrossRef]
  96. Bhachoo, J.; Beuming, T. Investigating protein peptide interactions using the schrödinger computational suite. In Modeling Peptide-Protein Interactions: Methods and Protocols; Schueler-Furman, O., London, N., Eds.; Methods in Molecular Biology; Springer: New York, NY, 2017; pp. 235–254. [Google Scholar] [CrossRef]
  97. Halder, S.K.; Ahmad, I.; Shathi, J.F.; Mim, M.M.; Hassan, M.R.; Jewel, M.J.I.; Dey, P.; Islam, M.S.; Patel, H.; Morshed, M.R.; Shakil, M.S.; Hossen, M.S. A Comprehensive study to unleash the putative inhibitors of serotype2 of dengue virus: insights from an in silico structure-based drug discovery. Mol. Biotechnol. 2022. [Google Scholar] [CrossRef]
Figure 4. 3D Simulation by molecular coupling between the DENV-2 protease NS2B/NS3 (PDB: 2FOM) versus chlorogenic acid using Chimera software V. 1.16.
Figure 4. 3D Simulation by molecular coupling between the DENV-2 protease NS2B/NS3 (PDB: 2FOM) versus chlorogenic acid using Chimera software V. 1.16.
Preprints 74381 g004
Figure 5. 3D Simulation by molecular coupling between the DENV-2 protease NS2B/NS3 (PDB: 2FOM) versus rosmarinic acid using Chimera software V. 1.16.
Figure 5. 3D Simulation by molecular coupling between the DENV-2 protease NS2B/NS3 (PDB: 2FOM) versus rosmarinic acid using Chimera software V. 1.16.
Preprints 74381 g005
Figure 6. 2D docked complexes of the molecular interaction between NS5B (PDB: 4E06) domain and chlorogenic acid (A), rutin (B), gallic acid (C), rosmarinic acid (D), and caffeic acid (E).
Figure 6. 2D docked complexes of the molecular interaction between NS5B (PDB: 4E06) domain and chlorogenic acid (A), rutin (B), gallic acid (C), rosmarinic acid (D), and caffeic acid (E).
Preprints 74381 g006
Figure 7. 3D Simulation by molecular coupling between the HCV protease NS5B (PDB: 4E06) versus rutin using Chimera software V. 1.16.
Figure 7. 3D Simulation by molecular coupling between the HCV protease NS5B (PDB: 4E06) versus rutin using Chimera software V. 1.16.
Preprints 74381 g007
Figure 8. 3D Simulation by molecular coupling between the HCV protease NS5B (PDB: 4E06) versus chlorogenic acid using Chimera software V. 1.16.
Figure 8. 3D Simulation by molecular coupling between the HCV protease NS5B (PDB: 4E06) versus chlorogenic acid using Chimera software V. 1.16.
Preprints 74381 g008
Figure 9. 3D Simulation by molecular coupling between the HCV protease NS5B (PDB: 4E06) versus rosmarinic acid using Chimera software V. 1.16.
Figure 9. 3D Simulation by molecular coupling between the HCV protease NS5B (PDB: 4E06) versus rosmarinic acid using Chimera software V. 1.16.
Preprints 74381 g009
Figure 10. 2D docked complexes of the molecular interaction between ICP7 (PDB: 4YXP) domain and chlorogenic acid (A), rutin (B), gallic acid (C), rosmarinic acid (D), and caffeic acid (E).
Figure 10. 2D docked complexes of the molecular interaction between ICP7 (PDB: 4YXP) domain and chlorogenic acid (A), rutin (B), gallic acid (C), rosmarinic acid (D), and caffeic acid (E).
Preprints 74381 g010
Figure 11. 3D Simulation by molecular coupling between the HSV-1 protease ICP7 (PDB: 4YXP) versus rutin using Chimera software V. 1.16.
Figure 11. 3D Simulation by molecular coupling between the HSV-1 protease ICP7 (PDB: 4YXP) versus rutin using Chimera software V. 1.16.
Preprints 74381 g011
Table 1. Taxonomical and ethnobotanical identification of medicinal plants sold by herbalists in markets of the city of Trujillo, La Libertad region.
Table 1. Taxonomical and ethnobotanical identification of medicinal plants sold by herbalists in markets of the city of Trujillo, La Libertad region.
Plant species Family Vernacular name 1 HUT code 2 Type of
viral affection
Part used 1 Way of
preparation 1
Way of use 1
Azadirachta indica A. Juss. Meliaceae Paraíso 60828 Mosquito-borne Le Inf A liter, three times a day
Caesalpinia spinosa
(Molina) Kuntze
Fabaceae Tara 60820 Dermatological, respiratory Po, WP Inf, De, Wa, Ba Gargle, two times a day
Wa, Ba: a time a day
Citrus limon (L.) Osbeck Rutaceae Limón 60821 Hepatic, dermatological, respiratory, COVID-19 Le, Fr Inf, Ba Time water
Clinopodium pulchellum (Kunth) Govaerts Lamiaceae Panizara 60830 Mosquito-borne WP Inf, De Time water
Cordia lutea Lam. Boraginaceae Overo 60823 Hepatic Fl Inf, De A liter, three times a day
Ocimum basilicum L. Lamiaceae Albahaca 60826 Respiratory, COVID-19, Mosquito-borne WP Inf, Ba A liter, three times a day
Ba: A time a day
Schinus molle L. Anacardaceae Molle 60836 Dermatological, respiratory, mosquito-borne Le, Se, WP Inf, Ba, Ru A liter, three times a day
Ba: three times a week
Taraxacum campylodes G. E. Haglund Asteraceae Diente de león 60838 Hepatic, respiratory, COVID-19 AP, WP Inf, De A liter as time water
1 Information provided by market herbalists through interviews; 2 Identified in the Herbarium Truxillense (HUT). Part Used: AP, aerial parts; Fl, flowers; Fr, fruits; Le, leaves; Po, pod; Se, seeds; WP, whole plant. Preparation: Ba, Baths; De, decoction; Inf, infusion; Ru, Rub; Wa, washes.
Table 2. Review of the antiviral activity reported from plant species.
Table 2. Review of the antiviral activity reported from plant species.
Virus Plant species Reference
Denge virus Azadirachta indica [28,29]
Clinopodium gracile [30]
Ocimum basilicum [31,32,33]
Schinus molle [34,35,36]
Herpes simplex virus Caesalpinia ferrea [37]
Caesalpinia pucherrima [38]
Citrus limon [39,40,41]
Schinus terebinthifolia [42,43,44]
Hepatitis virus Caesalpinia crista [45]
Caesalpinia sappan [46]
Citrus limon [22]
Cordia lutea [47,48]
Taraxacum officinale [49]
Table 3. Identification of potential antiviral compounds by LC-MS/MS.
Table 3. Identification of potential antiviral compounds by LC-MS/MS.
No Rt (Min) Chemical formula [M-H]-
(m/z)
Fragment ions
(m/z)
Identified
compound
Plant species
1 1.87 C7H6O5 169 125, 78, 69 Gallic acid Azadirachta indica
Caesalpinia spinosa
2 3.89 C16H18O9 353 191, 179 Chlorogenic acid Clinopodium pulchellum
Schinus molle
Taraxacum compylodes
3 5.97 C9H8O4 179 135, 107 Caffeic acid Azadirachta indica
Ocimun basilicum
Taraxacum compylodes
4 10.31 C27H30O16 609 301, 169 Rutin Azadirachta indica
Citrus limon
Clinopodium pulchellumCordia lutea
Schinus molle
5 13.91 C18H16O8 359 197, 179, 161 Rosmarinic acid Clinopodium pulchellum Ocimun basilicum
Table 4. Molecular docking values of the polyphenols with potential antiviral activity.
Table 4. Molecular docking values of the polyphenols with potential antiviral activity.
Pubchem ID Ligand Parameters DENV-2
(PDB: 2FOM)
HCV
(PDB: 4EO6)
HSV-1
(PDB: 4YXP)
1794427 Chlorogenic acid Score -8.6 -8.2 -6.8
H-Bonds 4 3 0
N. torsions 11 11 11
Bindig residues TRP83, LEU149 ASN142, GLU398,
ASN411
-
5280805 Rutin Score -7.1 -9.9 -7.8
H-Bonds 2 2 3
N. torsions 16 16 16
Bindig residues GLY153(2) ASP444, GLY557 ARG345, ARG393
0370 Gallic acid Score -5.7 -5.9 -5.6
H-Bonds 2 0 3
N. torsions 5 5 5
Bindig residues ALA164, ASN152 - ARG417, ARG435, ARG439
5281792 Rosmarinic acid Score -7.9 -7.9 -6.8
H-Bonds 2 2 1
N. torsions 12 12 12
Bindig residues ALA164, LEU149 ALA97, GLY449 GLU358
689043 Caffeic acid Score -6.2 -6.1 -5.7
H-Bonds 1 0 1
N. torsions 5 5 5
Bindig residues LEU149 - GLU351
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.
Copyright: This open access article is published under a Creative Commons CC BY 4.0 license, which permit the free download, distribution, and reuse, provided that the author and preprint are cited in any reuse.
Prerpints.org logo

Preprints.org is a free preprint server supported by MDPI in Basel, Switzerland.

Subscribe

Disclaimer

Terms of Use

Privacy Policy

Privacy Settings

© 2025 MDPI (Basel, Switzerland) unless otherwise stated